74 results on '"Shammas MA"'
Search Results
2. Telomeres, lifestyle, cancer, and aging.
- Author
-
Shammas MA and Shammas, Masood A
- Published
- 2011
- Full Text
- View/download PDF
3. Sulforaphane induces cell cycle arrest by protecting RB-E2F-1 complex in epithelial ovarian cancer cells
- Author
-
Morris Robert, Qazi Aamer M, Seward Shelly, Haider Mahdi, Pal Jagannath, Shah Jay, Chamala Sreedhar, Kumar Sanjeev, Bryant Christopher S, Semaan Assaad, Shammas Masood A, Steffes Christopher, Potti Ravindra B, Prasad Madhu, Weaver Donald W, and Batchu Ramesh B
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Sulforaphane (SFN), an isothiocyanate phytochemical present predominantly in cruciferous vegetables such as brussels sprout and broccoli, is considered a promising chemo-preventive agent against cancer. In-vitro exposure to SFN appears to result in the induction of apoptosis and cell-cycle arrest in a variety of tumor types. However, the molecular mechanisms leading to the inhibition of cell cycle progression by SFN are poorly understood in epithelial ovarian cancer cells (EOC). The aim of this study is to understand the signaling mechanisms through which SFN influences the cell growth and proliferation in EOC. Results SFN at concentrations of 5 - 20 μM induced a dose-dependent suppression of growth in cell lines MDAH 2774 and SkOV-3 with an IC50 of ~8 μM after a 3 day exposure. Combination treatment with chemotherapeutic agent, paclitaxel, resulted in additive growth suppression. SFN at ~8 μM decreased growth by 40% and 20% on day 1 in MDAH 2774 and SkOV-3, respectively. Cells treated with cytotoxic concentrations of SFN have reduced cell migration and increased apoptotic cell death via an increase in Bak/Bcl-2 ratio and cleavage of procaspase-9 and poly (ADP-ribose)-polymerase (PARP). Gene expression profile analysis of cell cycle regulated proteins demonstrated increased levels of tumor suppressor retinoblastoma protein (RB) and decreased levels of E2F-1 transcription factor. SFN treatment resulted in G1 cell cycle arrest through down modulation of RB phosphorylation and by protecting the RB-E2F-1 complex. Conclusions SFN induces growth arrest and apoptosis in EOC cells. Inhibition of retinoblastoma (RB) phosphorylation and reduction in levels of free E2F-1 appear to play an important role in EOC growth arrest.
- Published
- 2010
- Full Text
- View/download PDF
4. Ritonavir blocks AKT signaling, activates apoptosis and inhibits migration and invasion in ovarian cancer cells
- Author
-
Weaver Donald W, Steffes Christopher P, Pal Jagannath, Seward Shelly, Qazi Aamer, Bryant Christopher S, Chamala Sreedhar, Kumar Sanjeev, Morris Robert, Malone John M, Shammas Masood A, Prasad Madhu, and Batchu Ramesh B
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Ovarian cancer is the leading cause of mortality from gynecological malignancies, often undetectable in early stages. The difficulty of detecting the disease in its early stages and the propensity of ovarian cancer cells to develop resistance to known chemotherapeutic treatments dramatically decreases the 5-year survival rate. Chemotherapy with paclitaxel after surgery increases median survival only by 2 to 3 years in stage IV disease highlights the need for more effective drugs. The human immunodeficiency virus (HIV) infection is characterized by increased risk of several solid tumors due to its inherent nature of weakening of immune system. Recent observations point to a lower incidence of some cancers in patients treated with protease inhibitor (PI) cocktail treatment known as HAART (Highly Active Anti-Retroviral Therapy). Results Here we show that ritonavir, a HIV protease inhibitor effectively induced cell cycle arrest and apoptosis in ovarian cell lines MDH-2774 and SKOV-3 in a dose dependent manner. Over a 3 day period with 20 μM ritonavir resulted in the cell death of over 60% for MDAH-2774 compared with 55% in case of SKOV-3 cell line. Ritonavir caused G1 cell cycle arrest of the ovarian cancer cells, mediated by down modulating levels of RB phosphorylation and depleting the G1 cyclins, cyclin-dependent kinase and increasing their inhibitors as determined by gene profile analysis. Interestingly, the treatment of ritonavir decreased the amount of phosphorylated AKT in a dose-dependent manner. Furthermore, inhibition of AKT by specific siRNA synergistically increased the efficacy of the ritonavir-induced apoptosis. These results indicate that the addition of the AKT inhibitor may increase the therapeutic efficacy of ritonavir. Conclusion Our results demonstrate a potential use of ritonavir for ovarian cancer with additive effects in conjunction with conventional chemotherapeutic regimens. Since ritonavir is clinically approved for human use for HIV, drug repositioning for ovarian cancer could accelerate the process of traditional drug development. This would reduce risks, limit the costs and decrease the time needed to bring the drug from bench to bedside.
- Published
- 2009
- Full Text
- View/download PDF
5. Telomerase inhibition by siRNA causes senescence and apoptosis in Barrett's adenocarcinoma cells: mechanism and therapeutic potential
- Author
-
Batchu Ramesh B, Koley Hemanta, Shammas Masood A, Bertheau Robert C, Protopopov Alexei, Munshi Nikhil C, and Goyal Raj K
- Subjects
SiRNA, Telomerase, Telomere ,Barrett's esophageal adenocarcinoma ,Senescence, Apoptosis, Cancer treatment ,Cancer prevention, P73, P63 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background In cancer cells, telomerase induction helps maintain telomere length and thereby bypasses senescence and provides enhanced replicative potential. Chemical inhibitors of telomerase have been shown to reactivate telomere shortening and cause replicative senescence and apoptotic cell death of tumor cells while having little or no effect on normal diploid cells. Results We designed siRNAs against two different regions of telomerase gene and evaluated their effect on telomere length, proliferative potential, and gene expression in Barrett's adenocarcinoma SEG-1 cells. The mixture of siRNAs in nanomolar concentrations caused a loss of telomerase activity that appeared as early as day 1 and was essentially complete at day 3. Inhibition of telomerase activity was associated with marked reduction in median telomere length and complete loss of detectable telomeres in more than 50% of the treated cells. Telomere loss caused senescence in 40% and apoptosis in 86% of the treated cells. These responses appeared to be associated with activation of DNA sensor HR23B and subsequent activation of p53 homolog p73 and p63 and E2F1. Changes in these gene regulators were probably the source of observed up-regulation of cell cycle inhibitors, p16 and GADD45. Elevated transcript levels of FasL, Fas and caspase 8 that activate death receptors and CARD 9 that interacts with Bcl10 and NFKB to enhance mitochondrial translocation and activation of caspase 9 were also observed. Conclusion These studies show that telomerase siRNAs can cause effective suppression of telomerase and telomere shortening leading to both cell cycle arrest and apoptosis via mechanisms that include up-regulation of several genes involved in cell cycle arrest and apoptosis. Telomerase siRNAs may therefore be strong candidates for highly selective therapy for chemoprevention and treatment of Barrett's adenocarcinoma.
- Published
- 2005
- Full Text
- View/download PDF
6. Synthetic miR-34a mimics as a novel therapeutic agent for Multiple Myeloma: in vitro and in vivo evidence
- Author
-
Kenneth C. Anderson, Umberto Foresta, Vera Tomaino, Marco Rossi, Francesco Conforti, Manlio Ferrarini, Masood A. Shammas, Marta Lionetti, Massimo Negrini, Eugenio Morelli, Michele Caraglia, Annamaria Gulla, Pierfrancesco Tassone, Nicola Amodio, Pierosandro Tagliaferri, Nikhil C. Munshi, Maria Eugenia Gallo Cantafio, Antonino Neri, Maria Teresa Di Martino, Maria Rita Pitari, Emanuela Leone, Di Martino, Mt, Leone, E, Amodio, N, Foresta, U, Lionetti, M, Pitari, Mr, Gallo Cantafio, Me, Gullà, A, Conforti, F, Morelli, E, Tomaino, V, Rossi, M, Negrini, M, Ferrarini, M, Caraglia, Michele, Shammas, Ma, Munshi, Nc, Anderson, Kc, Neri, A, Tagliaferri, P, and Tassone, P.
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Stromal cell ,Apoptosis ,Mice, SCID ,Biology ,Transfection ,Article ,Cell Line ,Mice ,chemistry.chemical_compound ,Transduction, Genetic ,In vivo ,plasma cell leukemia ,microRNA ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Cell Proliferation ,Tumor microenvironment ,Cell growth ,Lentivirus ,Genetic Therapy ,In vitro ,Tumor Burden ,multiple myeloma ,MicroRNAs ,SCID-synth-hu model ,Oncology ,chemistry ,Cell culture ,miRNAs ,Cancer research ,RNA Interference ,miR-34a ,Growth inhibition ,Neoplasm Transplantation - Abstract
Purpose: Deregulated expression of miRNAs has been shown in multiple myeloma (MM). A promising strategy to achieve a therapeutic effect by targeting the miRNA regulatory network is to enforce the expression of miRNAs that act as tumor suppressor genes, such as miR-34a. Experimental Design: Here, we investigated the therapeutic potential of synthetic miR-34a against human MM cells in vitro and in vivo. Results: Either transient expression of miR-34a synthetic mimics or lentivirus-based miR-34a-stable enforced expression triggered growth inhibition and apoptosis in MM cells in vitro. Synthetic miR-34a downregulated canonic targets BCL2, CDK6, and NOTCH1 at both the mRNA and protein level. Lentiviral vector-transduced MM xenografts with constitutive miR-34a expression showed high growth inhibition in severe combined immunodeficient (SCID) mice. The anti-MM activity of lipidic-formulated miR-34a was further shown in vivo in two different experimental settings: (i) SCID mice bearing nontransduced MM xenografts; and (ii) SCID-synth-hu mice implanted with synthetic 3-dimensional scaffolds reconstituted with human bone marrow stromal cells and then engrafted with human MM cells. Relevant tumor growth inhibition and survival improvement were observed in mice bearing TP53-mutated MM xenografts treated with miR-34a mimics in the absence of systemic toxicity. Conclusions: Our findings provide a proof-of-principle that formulated synthetic miR-34a has therapeutic activity in preclinical models and support a framework for development of miR-34a–based treatment strategies in MM patients. Clin Cancer Res; 18(22); 6260–70. ©2012 AACR.
- Published
- 2012
7. Development of hyperdiploidy starts at an early age and takes a decade to complete.
- Author
-
Samur MK, Aktas Samur A, Shah P, Park J, Fulciniti M, Shammas MA, Corre J, Anderson KC, Parmigiani G, Avet-Loiseau H, and Munshi NC
- Abstract
Nearly half of multiple myeloma (MM) patients have hyperdiploidy (HMM) at diagnosis. Although HMM occurs early, the mutational processes before and after hyperdiploidy are still unclear. Here, we used 72 WGS samples from patients with HMM and identified pre and post HMM mutation to define the chronology of development of hyperdipoidy. A MM cell accumulated on median 0.56 mutations per mb pre-HMM and for every clonal pre-HMM mutation, 1.21 mutations accumulated post-HMM. This analysis using mutations before and after hyperdiploidy show that hyperdiploidy happens after somatic hypermutation, pre-hyperdipoidy muations are AID and age/Clock-like signature driven whereas post-hyperdiploidy mutations are from DNA damage and APOBEC. Interestingly, the first hyperdiploidy event occured within the first 3 decades of life and took a decade to complete. Copy number changes affecting chromosomes 15 and 19 occurred first. Finally, mutations pre initiating event affected chromosomes at different rates while post-initiating event mutational processes affect each chromosomes equally., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
8. A novel metric-based approach of scoring early host immune response from oro-nasopharyngeal swabs predicts COVID-19 outcome.
- Author
-
Rajput Y, Neral A, Sherwani N, Jain V, Sahu M, Paikra F, Kushwaha A, Sahu A, Lodhi H, Sundrani O, Panda RK, Jain V, Shammas MA, and Pal J
- Subjects
- Humans, Male, Female, Middle Aged, RNA, Viral genetics, Aged, Nasopharynx virology, Nasopharynx immunology, Adult, Biomarkers, Prospective Studies, Oropharynx virology, Oropharynx immunology, Prognosis, Adaptive Immunity, T-Lymphocytes immunology, COVID-19 immunology, COVID-19 diagnosis, COVID-19 virology, Interleukin-10 genetics, SARS-CoV-2 immunology, SARS-CoV-2 isolation & purification
- Abstract
Unpredictable fatal outcome of COVID-19 is attributed to dysregulated inflammation. Impaired early adaptive immune response leads to late-stage inflammatory outcome. The purpose of this study was to develop biomarkers for early detection of host immune impairment at first diagnosis from leftover RNA samples, which may in turn identify high risk patients. Leftover RNA samples of COVID-19 patients at first diagnosis were stored. Following prospective follow-up, the samples were shorted and categorized into outcome groups. Impaired adaptive T cell response (severity score) and Impaired IL-10 response (undetectable IL-10 in the presence of high expression of a representative interferon response gene) were determined by RT-PCR based assay. We demonstrate that a T cell response based 'severity score' comprising rational combination of Ct values of a target genes' signature can predict high risk noncomorbid potentially critical COVID-19 patients with a sensitivity of 91% (95% CI 58.7-99.8) and specificity of 92.6% (95% CI 75.7-99) (AUC:0.88). Although inclusion of comorbid patients reduced sensitivity to 77% (95% CI 54.6-92.2), the specificity was still 94% (95% CI 79.8-99.3) (AUC:0.82). The same for 'impaired IL-10 response' were little lower to predict high risk noncomorbid patients 64.2% (95% CI 35.1-87.2) and 82% (95% CI 65.5-93.2) respectively. Inclusion of comorbid patients drastically reduce sensitivity and specificity51.6% (95% CI 33.1-69.8) and 80.5% (95% CI 64.0-91.8) respectively. As best of our knowledge this is the first demonstration of a metric-based approach showing the 'severity score' as an indicator of early adoptive immune response, could be used as predictor of severe COVID-19 outcome at the time of first diagnosis using the same leftover swab RNA. The work flow could reduce expenditure and reporting time of the prognostic test for an earliest clinical decision ensuring possibility of early rational management., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Retraction: Effects of Oligonucleotide N3'→P5' Thio-phosphoramidate (GRN163) Targeting Telomerase RNA in Human Multiple Myeloma Cells.
- Author
-
Akiyama M, Hideshima T, Shammas MA, Hayashi T, Hamasaki M, Tai YT, Richardson P, Gryaznov S, Munshi NC, and Anderson KC
- Published
- 2024
- Full Text
- View/download PDF
10. ABL1 kinase plays an important role in spontaneous and chemotherapy-induced genomic instability in multiple myeloma.
- Author
-
Kumar S, Talluri S, Zhao J, Liao C, Potluri LB, Buon L, Mu S, Shi J, Chakraborty C, Tai YT, Samur MK, Munshi NC, and Shammas MA
- Subjects
- Humans, Melphalan pharmacology, Genomic Instability, Pyrimidines pharmacology, Pyrimidines therapeutic use, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Abstract: Genomic instability contributes to cancer progression and is at least partly due to dysregulated homologous recombination (HR). Here, we show that an elevated level of ABL1 kinase overactivates the HR pathway and causes genomic instability in multiple myeloma (MM) cells. Inhibiting ABL1 with either short hairpin RNA or a pharmacological inhibitor (nilotinib) inhibits HR activity, reduces genomic instability, and slows MM cell growth. Moreover, inhibiting ABL1 reduces the HR activity and genomic instability caused by melphalan, a chemotherapeutic agent used in MM treatment, and increases melphalan's efficacy and cytotoxicity in vivo in a subcutaneous tumor model. In these tumors, nilotinib inhibits endogenous as well as melphalan-induced HR activity. These data demonstrate that inhibiting ABL1 using the clinically approved drug nilotinib reduces MM cell growth, reduces genomic instability in live cell fraction, increases the cytotoxicity of melphalan (and similar chemotherapeutic agents), and can potentially prevent or delay progression in patients with MM.
- Published
- 2024
- Full Text
- View/download PDF
11. Elevated APE1 Dysregulates Homologous Recombination and Cell Cycle Driving Genomic Evolution, Tumorigenesis, and Chemoresistance in Esophageal Adenocarcinoma.
- Author
-
Kumar S, Zhao J, Talluri S, Buon L, Mu S, Potluri LB, Liao C, Shi J, Chakraborty C, Gonzalez GB, Tai YT, Patel J, Pal J, Mashimo H, Samur MK, Munshi NC, and Shammas MA
- Subjects
- Male, Animals, Mice, Humans, In Situ Hybridization, Fluorescence, Cell Line, Tumor, Carcinogenesis genetics, Cell Transformation, Neoplastic genetics, Homologous Recombination, Cell Cycle, Genomic Instability, Genomics, Chromosomal Instability genetics, Deoxyribonucleases genetics, Evolution, Molecular, Drug Resistance, Neoplasm genetics, Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma metabolism
- Abstract
Background & Aims: The purpose of this study was to identify drivers of genomic evolution in esophageal adenocarcinoma (EAC) and other solid tumors., Methods: An integrated genomics strategy was used to identify deoxyribonucleases correlating with genomic instability (as assessed from total copy number events in each patient) in 6 cancers. Apurinic/apyrimidinic nuclease 1 (APE1), identified as the top gene in functional screens, was either suppressed in cancer cell lines or overexpressed in normal esophageal cells and the impact on genome stability and growth was monitored in vitro and in vivo. The impact on DNA and chromosomal instability was monitored using multiple approaches, including investigation of micronuclei, acquisition of single nucleotide polymorphisms, whole genome sequencing, and/or multicolor fluorescence in situ hybridization., Results: Expression of 4 deoxyribonucleases correlated with genomic instability in 6 human cancers. Functional screens of these genes identified APE1 as the top candidate for further evaluation. APE1 suppression in EAC, breast, lung, and prostate cancer cell lines caused cell cycle arrest; impaired growth and increased cytotoxicity of cisplatin in all cell lines and types and in a mouse model of EAC; and inhibition of homologous recombination and spontaneous and chemotherapy-induced genomic instability. APE1 overexpression in normal cells caused a massive chromosomal instability, leading to their oncogenic transformation. Evaluation of these cells by means of whole genome sequencing demonstrated the acquisition of changes throughout the genome and identified homologous recombination as the top mutational process., Conclusions: Elevated APE1 dysregulates homologous recombination and cell cycle, contributing to genomic instability, tumorigenesis, and chemoresistance, and its inhibitors have the potential to target these processes in EAC and possibly other cancers., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. High-dose melphalan treatment significantly increases mutational burden at relapse in multiple myeloma.
- Author
-
Samur MK, Roncador M, Aktas Samur A, Fulciniti M, Bazarbachi AH, Szalat R, Shammas MA, Sperling AS, Richardson PG, Magrangeas F, Minvielle S, Perrot A, Corre J, Moreau P, Thakurta A, Parmigiani G, Anderson KC, Avet-Loiseau H, and Munshi NC
- Subjects
- Humans, Melphalan therapeutic use, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Bortezomib therapeutic use, Lenalidomide therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chronic Disease, Transplantation, Autologous, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma diagnosis
- Abstract
High-dose melphalan (HDM) improves progression-free survival in multiple myeloma (MM), yet melphalan is a DNA-damaging alkylating agent; therefore, we assessed its mutational effect on surviving myeloma cells by analyzing paired MM samples collected at diagnosis and relapse in the IFM 2009 study. We performed deep whole-genome sequencing on samples from 68 patients, 43 of whom were treated with RVD (lenalidomide, bortezomib, and dexamethasone) and 25 with RVD + HDM. Although the number of mutations was similar at diagnosis in both groups (7137 vs 7230; P = .67), the HDM group had significantly more mutations at relapse (9242 vs 13 383, P = .005). No change in the frequency of copy number alterations or structural variants was observed. The newly acquired mutations were typically associated with DNA damage and double-stranded breaks and were predominantly on the transcribed strand. A machine learning model, using this unique pattern, predicted patients who would receive HDM with high sensitivity, specificity, and positive prediction value. Clonal evolution analysis showed that all patients treated with HDM had clonal selection, whereas a static progression was observed with RVD. A significantly higher percentage of mutations were subclonal in the HDM cohort. Intriguingly, patients treated with HDM who achieved complete remission (CR) had significantly more mutations at relapse yet had similar survival rates as those treated with RVD who achieved CR. This similarity could have been due to HDM relapse samples having significantly more neoantigens. Overall, our study identifies increased genomic changes associated with HDM and provides rationale to further understand clonal complexity., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
13. A MIR17HG-derived long noncoding RNA provides an essential chromatin scaffold for protein interaction and myeloma growth.
- Author
-
Morelli E, Fulciniti M, Samur MK, Ribeiro CF, Wert-Lamas L, Henninger JE, Gullà A, Aktas-Samur A, Todoerti K, Talluri S, Park WD, Federico C, Scionti F, Amodio N, Bianchi G, Johnstone M, Liu N, Gramegna D, Maisano D, Russo NA, Lin C, Tai YT, Neri A, Chauhan D, Hideshima T, Shammas MA, Tassone P, Gryaznov S, Young RA, Anderson KC, Novina CD, Loda M, and Munshi NC
- Subjects
- Humans, Animals, Mice, Chromatin, Cell Proliferation, Gene Expression Regulation, Neoplastic, RNA, Long Noncoding genetics, Multiple Myeloma genetics, MicroRNAs metabolism
- Abstract
Long noncoding RNAs (lncRNAs) can drive tumorigenesis and are susceptible to therapeutic intervention. Here, we used a large-scale CRISPR interference viability screen to interrogate cell-growth dependency to lncRNA genes in multiple myeloma (MM) and identified a prominent role for the miR-17-92 cluster host gene (MIR17HG). We show that an MIR17HG-derived lncRNA, named lnc-17-92, is the main mediator of cell-growth dependency acting in a microRNA- and DROSHA-independent manner. Lnc-17-92 provides a chromatin scaffold for the functional interaction between c-MYC and WDR82, thus promoting the expression of ACACA, which encodes the rate-limiting enzyme of de novo lipogenesis acetyl-coA carboxylase 1. Targeting MIR17HG pre-RNA with clinically applicable antisense molecules disrupts the transcriptional and functional activities of lnc-17-92, causing potent antitumor effects both in vitro and in vivo in 3 preclinical animal models, including a clinically relevant patient-derived xenograft NSG mouse model. This study establishes a novel oncogenic function of MIR17HG and provides potent inhibitors for translation to clinical trials., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
14. RAD51 Is Implicated in DNA Damage, Chemoresistance and Immune Dysregulation in Solid Tumors.
- Author
-
Liao C, Talluri S, Zhao J, Mu S, Kumar S, Shi J, Buon L, Munshi NC, and Shammas MA
- Abstract
Background: In normal cells, homologous recombination (HR) is tightly regulated and plays an important role in the maintenance of genomic integrity and stability through precise repair of DNA damage. RAD51 is a recombinase that mediates homologous base pairing and strand exchange during DNA repair by HR. Our previous data in multiple myeloma and esophageal adenocarcinoma (EAC) show that dysregulated HR mediates genomic instability. Purpose of this study was to investigate role of HR in genomic instability, chemoresistance and immune dysregulation in solid tumors including colon and breast cancers., Methods: The GEO dataset were used to investigate correlation of RAD51 expression with patient survival and expression of various immune markers in EAC, breast and colorectal cancers. RAD51 was inhibited in cancer cell lines using shRNAs and a small molecule inhibitor. HR activity was evaluated using a plasmid-based assay, DNA breaks assessed by evaluating expression of γ-H2AX (a marker of DNA breaks) and p-RPA32 (a marker of DNA end resection) using Western blotting. Genomic instability was monitored by investigating micronuclei (a marker of genomic instability). Impact of RAD51 inhibitor and/or a DNA-damaging agent was assessed on viability and apoptosis in EAC, breast and colon cancer cell lines in vitro and in a subcutaneous tumor model of EAC. Impact of RAD51 inhibitor on expression profile was monitored by RNA sequencing., Results: Elevated RAD51 expression correlated with poor survival of EAC, breast and colon cancer patients. RAD51 knockdown in cancer cell lines inhibited DNA end resection and strand exchange activity (key steps in the initiation of HR) as well as spontaneous DNA breaks, whereas its overexpression increased DNA breaks and genomic instability. Treatment of EAC, colon and breast cancer cell lines with a small molecule inhibitor of RAD51 inhibited DNA breaking agent-induced DNA breaks and genomic instability. RAD51 inhibitor potentiated cytotoxicity of DNA breaking agent in all cancer cell types tested in vitro as well as in a subcutaneous model of EAC. Evaluation by RNA sequencing demonstrated that DNA repair and cell cycle related pathways were induced by DNA breaking agent whereas their induction either prevented or reversed by RAD51 inhibitor. In addition, immune-related pathways such as PD-1 and Interferon Signaling were also induced by DNA breaking agent whereas their induction prevented by RAD51 inhibitor. Consistent with these observations, elevated RAD51 expression also correlated with that of genes involved in inflammation and other immune surveillance., Conclusions: Elevated expression of RAD51 and associated HR activity is involved in spontaneous and DNA damaging agent-induced DNA breaks and genomic instability thus contributing to chemoresistance, immune dysregulation and poor prognosis in cancer. Therefore, inhibitors of RAD51 have great potential as therapeutic agents for EAC, colon, breast and probably other solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
15. A standalone approach to utilize telomere length measurement as a surveillance tool in oral leukoplakia.
- Author
-
Pal J, Rajput Y, Shrivastava S, Gahine R, Mungutwar V, Barardiya T, Chandrakar A, Ramakrishna PP, Mishra SS, Banjara H, Choudhary V, Patra PK, and Shammas MA
- Subjects
- Female, Humans, Leukocytes, Mononuclear metabolism, Leukoplakia, Oral diagnosis, Leukoplakia, Oral genetics, Leukoplakia, Oral metabolism, Male, Telomere metabolism, Telomere pathology, Carcinoma, Squamous Cell diagnosis, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms, Mouth Neoplasms genetics
- Abstract
Oral squamous cell carcinoma (OSCC) is often preceded by a white patch on a surface of the mouth, called oral leukoplakia (OL). As accelerated telomere length (TL) shortening in dividing epithelial cells may lead to oncogenic transformation, telomere length measurement could serve as a predictive biomarker in OL. However, due to high variability and lack of a universal reference, there has been a limited translational application. Here, we describe an approach of evaluating TL using paired peripheral blood mononuclear cells (PBMC) as an internal reference and demonstrate its translational relevance. Oral brush biopsy and paired venous blood were collected from 50 male OL patients and 44 male healthy controls (HC). Relative TL was measured by quantitative PCR. TL of each OL or healthy sample was normalized to the paired PBMC sample (TL ratio). In OL patients, the mean TL ratio was significantly smaller not only in the patch but also in distal normal oral tissue, relative to healthy controls without a high-risk oral habit. Dysplasia was frequently associated with a subgroup that showed a normal TL ratio at the patch but significantly smaller TL ratio at a paired normal distal site. Our data suggest that evaluation of TL attrition using a paired PBMC sample eliminates the requirement of external reference DNA, makes data universally comparable and provides a useful marker to define high-risk OL groups for follow-up programs. Larger studies will further validate the approach and its broader application in other premalignant conditions., (© 2021 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2022
- Full Text
- View/download PDF
16. Dysregulated APOBEC3G causes DNA damage and promotes genomic instability in multiple myeloma.
- Author
-
Talluri S, Samur MK, Buon L, Kumar S, Potluri LB, Shi J, Prabhala RH, Shammas MA, and Munshi NC
- Subjects
- APOBEC-3G Deaminase genetics, Cell Line, Tumor, Humans, Multiple Myeloma genetics, Neoplasm Proteins genetics, APOBEC-3G Deaminase metabolism, DNA Damage, Genomic Instability, Multiple Myeloma enzymology, Mutation, Neoplasm Proteins metabolism
- Abstract
Multiple myeloma (MM) is a heterogeneous disease characterized by significant genomic instability. Recently, a causal role for the AID/APOBEC deaminases in inducing somatic mutations in myeloma has been reported. We have identified APOBEC/AID as a prominent mutational signature at diagnosis with further increase at relapse in MM. In this study, we identified upregulation of several members of APOBEC3 family (A3A, A3B, A3C, and A3G) with A3G, as one of the most expressed APOBECs. We investigated the role of APOBEC3G in MM and observed that A3G expression and APOBEC deaminase activity is elevated in myeloma cell lines and patient samples. Loss-of and gain-of function studies demonstrated that APOBEC3G significantly contributes to increase in DNA damage (abasic sites and DNA breaks) in MM cells. Evaluation of the impact on genome stability, using SNP arrays and whole genome sequencing, indicated that elevated APOBEC3G contributes to ongoing acquisition of both the copy number and mutational changes in MM cells over time. Elevated APOBEC3G also contributed to increased homologous recombination activity, a mechanism that can utilize increased DNA breaks to mediate genomic rearrangements in cancer cells. These data identify APOBEC3G as a novel gene impacting genomic evolution and underlying mechanisms in MM., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)
- Published
- 2021
- Full Text
- View/download PDF
17. Integrated genomics and comprehensive validation reveal drivers of genomic evolution in esophageal adenocarcinoma.
- Author
-
Kumar S, Buon L, Talluri S, Roncador M, Liao C, Zhao J, Shi J, Chakraborty C, Gonzalez G, Tai YT, Prabhala R, Samur MK, Munshi NC, and Shammas MA
- Subjects
- Adenocarcinoma genetics, Animals, Apoptosis, Biomarkers, Tumor genetics, Cell Proliferation, Esophageal Neoplasms genetics, Female, Genomic Instability, Humans, Mice, Mice, Inbred BALB C, Mice, SCID, Prognosis, Survival Rate, Tumor Cells, Cultured, Whole Genome Sequencing, Xenograft Model Antitumor Assays, Adenocarcinoma pathology, Biomarkers, Tumor metabolism, Esophageal Neoplasms pathology, Evolution, Molecular, Gene Expression Regulation, Neoplastic, Genomics methods, Mutation
- Abstract
Esophageal adenocarcinoma (EAC) is associated with a marked genomic instability, which underlies disease progression and development of resistance to treatment. In this study, we used an integrated genomics approach to identify a genomic instability signature. Here we show that elevated expression of this signature correlates with poor survival in EAC as well as three other cancers. Knockout and overexpression screens establish the relevance of these genes to genomic instability. Indepth evaluation of three genes (TTK, TPX2 and RAD54B) confirms their role in genomic instability and tumor growth. Mutational signatures identified by whole genome sequencing and functional studies demonstrate that DNA damage and homologous recombination are common mechanisms of genomic instability induced by these genes. Our data suggest that the inhibitors of TTK and possibly other genes identified in this study have potential to inhibit/reduce growth and spontaneous as well as chemotherapy-induced genomic instability in EAC and possibly other cancers.
- Published
- 2021
- Full Text
- View/download PDF
18. RAD51 Inhibitor Reverses Etoposide-Induced Genomic Toxicity and Instability in Esophageal Adenocarcinoma Cells.
- Author
-
Liao C, Zhao J, Kumar S, Chakraborty C, Talluri S, Munshi NC, and Shammas MA
- Abstract
Aim: In normal cells, homologous recombination (HR) is strictly regulated and precise and plays an important role in preserving genomic integrity by accurately repairing DNA damage. RAD51 is the recombinase which mediates homologous base pairing and strand exchange during DNA repair by HR. We have previously reported that HR is spontaneously elevated (or dysregulated) in esophageal adenocarcinoma (EAC) and contributes to ongoing genomic changes and instability. The purpose of this study was to evaluate the impact of RAD51 inhibitor on genomic toxicity caused by etoposide, a chemotherapeutic agent., Methods: EAC cell lines (FLO-1 and OE19) were cultured in the presence of RAD51 inhibitor and/or etoposide, and impact on cell viability, apoptosis and genomic integrity/stability investigated. Genomic integrity/stability was monitored by evaluating cells for γ-H2AX (a marker for DNA breaks), phosphorylated RPA32 (a marker of DNA end resection which is a distinct step in the initiation of HR) and micronuclei (a marker of genomic instability)., Results: Treatment with etoposide, a chemotherapeutic agent, was associated with marked genomic toxicity (as evident from increase in DNA breaks) and genomic instability in both EAC cell lines. Consistently, the treatment was also associated with apoptotic cell death. A small molecule inhibitor of RAD51 increased cytotoxicity while reducing genomic toxicity and instability caused by etoposide, in both EAC cell lines., Conclusion: RAD51 inhibitors have potential to increase cytotoxicity while reducing harmful genomic impact of chemotherapy., Competing Interests: Conflict of Interest The authors declare that they have no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
19. Amplification and overexpression of E2 ubiquitin conjugase UBE2T promotes homologous recombination in multiple myeloma.
- Author
-
Alagpulinsa DA, Kumar S, Talluri S, Nanjappa P, Buon L, Chakraborty C, Samur MK, Szalat R, Shammas MA, and Munshi NC
- Subjects
- Humans, Multiple Myeloma pathology, Homologous Recombination genetics, Multiple Myeloma genetics, Ubiquitin metabolism, Ubiquitin-Conjugating Enzymes metabolism
- Published
- 2019
- Full Text
- View/download PDF
20. MUS81 Participates in the Progression of Serous Ovarian Cancer Associated With Dysfunctional DNA Repair System.
- Author
-
Lu R, Xie S, Wang Y, Zheng H, Zhang H, Deng M, Shi W, Zhong A, Chen M, Zhang M, Xu X, Shammas MA, and Guo L
- Abstract
Objective: Methyl methanesulfonate ultraviolet sensitive gene clone 81 (MUS81) is a structure-specific endonuclease that plays a pivotal role in the DNA repair system of cancer cells. In this study, we aim to elucidate the potential association between the dysfunction of MUS81 and the progression of Serous Ovarian Cancer (SOC). Methods: To investigate the association between MUS81 and prognosis of SOC, immunohistochemistry technology and qPCR were used to analyze the level of MUS81 expression, and transcriptional profile analysis and protein interaction screening chip were used to explore the MUS81 related signal pathways. Random amplified polymorphic DNA (RAPD) analysis, immunofluorescence and comet assays were further performed to evaluate genomic instability and DNA damage status of transduced SOC cells. Experiments both in vitro and in vivo were conducted to verify the impact of MUS81 silencing on chemotherapeutic drug sensitivity of SOC. Results: The overexpression of MUS81 in SOC tissues was related to poor clinical outcomes. The transcriptional chip data showed that MUS81 was involved in multiple pathways associated with DNA repair. Deficiency of MUS81 intensified the genome instability of SOC cells, promoted the emergence of DSBs and restrained the formation of RAD51 foci in SOC cells with exposure to UV. Furthermore, downregulation of MUS81 enhanced the sensitivity to Camptothecin and Olaparib in SOC cell lines and xenograft model. Conclusions: MUS81 is involved in the progression of SOC and inhibition of MUS81 could augment the susceptibility to chemotherapeutic agents. MUS81 might represent a novel molecular target for SOC chemotherapy., (Copyright © 2019 Lu, Xie, Wang, Zheng, Zhang, Deng, Shi, Zhong, Chen, Zhang, Xu, Shammas and Guo.)
- Published
- 2019
- Full Text
- View/download PDF
21. The roles of homologous recombination and the immune system in the genomic evolution of cancer.
- Author
-
Nandi B, Talluri S, Kumar S, Yenumula C, Gold JS, Prabhala R, Munshi NC, and Shammas MA
- Abstract
A variety of factors, whether extracellular (mutagens/carcinogens and viruses in the environment, chronic inflammation and radiation associated with the environment and/or electronic devices/machines) and/or intracellular (oxidative metabolites of food, oxidative stress due to inflammation, acid production, replication stress, DNA replication/repair errors, and certain hormones, cytokines, growth factors), pose a constant threat to the genomic integrity of a living cell. However, in the normal cellular environment multiple biological pathways including DNA repair, cell cycle, apoptosis and the immune system work in a precise, regulated (tightly controlled), timely and concerted manner to ensure genomic integrity, stability and proper functioning of a cell. If damage to DNA takes place, it is efficiently and accurately repaired by the DNA repair systems. Homologous recombination (HR) which utilizes either a homologous chromosome (in G1 phase) or a sister chromatid (in G2) as a template to repair the damage, is known to be the most precise repair system. HR in G2 which utilizes a sister chromatid as a template is also called an error free repair system. If DNA damage in a cell is so extensive that it overwhelms the repair system/s, the cell is eliminated by apoptosis. Thus, multiple pathways ensure that genome of a cell is intact and stable. However, constant exposure to DNA damage and/or dysregulation of DNA repair mechanism/s poses a risk of mutation and cancer. Oncogenesis, which seems to be a multistep process, is associated with acquisition of a number of genomic changes that enable a normal cell to progress from benign to malignant transformation. Transformed/cancer cells are recognized and killed by the immune system. However, the ongoing acquisition of new genomic changes enables cancer cells to survive/escape immune attack, evolve into a more aggressive phenotype, and eventually develop resistance to therapy. Although DNA repair (especially the HR) and the immune system play unique roles in preserving genomic integrity of a cell, they can also contribute to DNA damage, genomic instability and oncogenesis. The purpose of this article is to highlight the roles of DNA repair (especially HR) and the immune system in genomic evolution, with special focus on gastrointestinal cancer.
- Published
- 2019
- Full Text
- View/download PDF
22. Dual PAK4-NAMPT Inhibition Impacts Growth and Survival, and Increases Sensitivity to DNA-Damaging Agents in Waldenström Macroglobulinemia.
- Author
-
Li N, Lopez MA, Linares M, Kumar S, Oliva S, Martinez-Lopez J, Xu L, Xu Y, Perini T, Senapedis W, Baloglu E, Shammas MA, Hunter Z, Anderson KC, Treon SP, Munshi NC, and Fulciniti M
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Cytokines antagonists & inhibitors, DNA Breaks, Double-Stranded drug effects, DNA Damage drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Nicotinamide Phosphoribosyltransferase antagonists & inhibitors, Rad51 Recombinase genetics, Waldenstrom Macroglobulinemia genetics, Waldenstrom Macroglobulinemia pathology, p21-Activated Kinases antagonists & inhibitors, Acrylamides pharmacology, Aminopyridines pharmacology, Cytokines genetics, Nicotinamide Phosphoribosyltransferase genetics, Waldenstrom Macroglobulinemia drug therapy, p21-Activated Kinases genetics
- Abstract
Purpose: p21-activated kinase 4 (PAK4) plays a significant biological and functional role in a number of malignancies, including multiple myeloma (MM). On the basis of our promising findings in MM, we here characterize PAK4 expression and role in WM cells, as well effect of dual PAK4-NAMPT inhibitor (KPT-9274) against WM cell growth and viability., Experimental Design: We have analyzed mRNA and protein expression levels of PAK4 in WM cells, and used loss-of-function approach to investigate its contribution to WM cell viability. We have further tested the in vitro and in vivo effect of KPT-9274 against WM cell growth and viability., Results: We report here high-level expression and functional role of PAK4 in WM, as demonstrated by shRNA-mediated knockdown; and significant impact of KPT-9274 on WM cell growth and viability. The growth inhibitory effect of KPT-9274 was associated with decreased PAK4 expression and NAMPT activity, as well as induction of apoptosis. Interestingly, in WM cell lines treated with KPT-9274, we detected a significant impact on DNA damage and repair genes. Moreover, we observed that apart from inducing DNA damage, KPT-9274 specifically decreased RAD51 and the double-strand break repair by the homologous recombination pathway. As a result, when combined with a DNA alkylating agents bendamustine and melphalan, KPT-9274 provided a synergistic inhibition of cell viability in WM cell lines and primary patient WM cells in vitro and in vivo ., Conclusions: These results support the clinical investigation of KPT-9274 in combination with DNA-damaging agent for treatment of WM., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
23. Role of apurinic/apyrimidinic nucleases in the regulation of homologous recombination in myeloma: mechanisms and translational significance.
- Author
-
Kumar S, Talluri S, Pal J, Yuan X, Lu R, Nanjappa P, Samur MK, Munshi NC, and Shammas MA
- Subjects
- Antineoplastic Agents, Alkylating pharmacology, Cell Line, Tumor, DNA Damage, DNA Repair, DNA-(Apurinic or Apyrimidinic Site) Lyase genetics, Drug Resistance, Neoplasm genetics, Endonucleases, Gene Expression Regulation, Neoplastic, Genomic Instability, Humans, Melphalan pharmacology, Micronuclei, Chromosome-Defective, Multifunctional Enzymes, Protein Interaction Mapping, Protein Interaction Maps, Rad51 Recombinase genetics, Transcription, Genetic, Translational Research, Biomedical, DNA-(Apurinic or Apyrimidinic Site) Lyase metabolism, Homologous Recombination, Multiple Myeloma genetics, Multiple Myeloma metabolism
- Abstract
We have previously reported that homologous recombination (HR) is dysregulated in multiple myeloma (MM) and contributes to genomic instability and development of drug resistance. We now demonstrate that base excision repair (BER) associated apurinic/apyrimidinic (AP) nucleases (APEX1 and APEX2) contribute to regulation of HR in MM cells. Transgenic as well as chemical inhibition of APEX1 and/or APEX2 inhibits HR activity in MM cells, whereas the overexpression of either nuclease in normal human cells, increases HR activity. Regulation of HR by AP nucleases could be attributed, at least in part, to their ability to regulate recombinase (RAD51) expression. We also show that both nucleases interact with major HR regulators and that APEX1 is involved in P73-mediated regulation of RAD51 expression in MM cells. Consistent with the role in HR, we also show that AP-knockdown or treatment with inhibitor of AP nuclease activity increases sensitivity of MM cells to melphalan and PARP inhibitor. Importantly, although inhibition of AP nuclease activity increases cytotoxicity, it reduces genomic instability caused by melphalan. In summary, we show that APEX1 and APEX2, major BER proteins, also contribute to regulation of HR in MM. These data provide basis for potential use of AP nuclease inhibitors in combination with chemotherapeutics such as melphalan for synergistic cytotoxicity in MM.
- Published
- 2018
- Full Text
- View/download PDF
24. Genomic patterns of progression in smoldering multiple myeloma.
- Author
-
Bolli N, Maura F, Minvielle S, Gloznik D, Szalat R, Fullam A, Martincorena I, Dawson KJ, Samur MK, Zamora J, Tarpey P, Davies H, Fulciniti M, Shammas MA, Tai YT, Magrangeas F, Moreau P, Corradini P, Anderson K, Alexandrov L, Wedge DC, Avet-Loiseau H, Campbell P, and Munshi N
- Subjects
- Aged, Databases, Genetic, Disease Progression, Female, Gene Expression Profiling, Genomics, Humans, Male, Middle Aged, Multiple Myeloma genetics, Multiple Myeloma pathology, Mutation genetics, Risk Factors, Smoldering Multiple Myeloma pathology, Gene Expression Regulation, Neoplastic, Smoldering Multiple Myeloma genetics
- Abstract
We analyzed whole genomes of unique paired samples from smoldering multiple myeloma (SMM) patients progressing to multiple myeloma (MM). We report that the genomic landscape, including mutational profile and structural rearrangements at the smoldering stage is very similar to MM. Paired sample analysis shows two different patterns of progression: a "static progression model", where the subclonal architecture is retained as the disease progressed to MM suggesting that progression solely reflects the time needed to accumulate a sufficient disease burden; and a "spontaneous evolution model", where a change in the subclonal composition is observed. We also observe that activation-induced cytidine deaminase plays a major role in shaping the mutational landscape of early subclinical phases, while progression is driven by APOBEC cytidine deaminases. These results provide a unique insight into myelomagenesis with potential implications for the definition of smoldering disease and timing of treatment initiation.
- Published
- 2018
- Full Text
- View/download PDF
25. Phosphatidylserine-exposing blood cells and microparticles induce procoagulant activity in non-valvular atrial fibrillation.
- Author
-
Wang L, Bi Y, Yu M, Li T, Tong D, Yang X, Zhang C, Guo L, Wang C, Kou Y, Dong Z, Novakovic VA, Tian Y, Kou J, Shammas MA, and Shi J
- Subjects
- Aged, Atrial Fibrillation physiopathology, Blood Cells chemistry, Blood Coagulation Tests methods, Cell-Derived Microparticles chemistry, Echocardiography methods, Electrocardiography methods, Female, Humans, Male, Microscopy, Confocal methods, Middle Aged, Phosphatidylserines analysis, Atrial Fibrillation blood, Atrial Fibrillation diagnostic imaging, Blood Cells metabolism, Blood Coagulation physiology, Cell-Derived Microparticles metabolism, Phosphatidylserines metabolism
- Abstract
Background: The definitive role of phosphatidylserine (PS) in the prothrombotic state of non-valvular atrial fibrillation (NVAF) remains unclear. Our objectives were to study the PS exposure on blood cells and microparticles (MPs) in NVAF, and evaluate their procoagulant activity (PCA)., Methods: NVAF patients without (n = 60) and with left atrial thrombi (n = 18) and controls (n = 36) were included in our study. Exposed PS was analyzed with flow cytometry and confocal microscopy. PCA was evaluated using clotting time, factor Xa (FXa), thrombin and fibrin formation., Results: PS
+ blood cells and MPs were significantly higher in NVAF patients without and with left atrial thrombi (both P < 0.01) than in controls. Patients with left atrial thrombi showed increased PS+ platelets, neutrophils, erythrocytes and MPs compared with patients without thrombi (all P < 0.05). Moreover, in patients with left atrial thrombi, MPs primarily originated from platelets (56.1%) followed by leukocytes (21.9%, including MPs from neutrophils, monocytes and lymphocytes), erythrocytes (12.2%) and endothelial cells (8.9%). Additionally, PS+ blood cells and MPs contributed to markedly shortened coagulation time and dramatically increased FXa/thrombin/fibrin (all P < 0.001) generation in both NVAF groups. Furthermore, blockade of exposed PS on blood cells and MPs with lactadherin inhibited PCA by approximately 80%. Lastly, we found that the amount of PS+ platelets and MPs was positively correlated with thrombus diameter (all p < 0.005)., Conclusions: Our results suggest that exposed PS on blood cells and MPs play a procoagulant role in NVAF patients. Blockade of PS prior to thrombus formation might be a novel therapeutic approach in these patients., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
26. Nucleotide excision repair is a potential therapeutic target in multiple myeloma.
- Author
-
Szalat R, Samur MK, Fulciniti M, Lopez M, Nanjappa P, Cleynen A, Wen K, Kumar S, Perini T, Calkins AS, Reznichenko E, Chauhan D, Tai YT, Shammas MA, Anderson KC, Fermand JP, Arnulf B, Avet-Loiseau H, Lazaro JB, and Munshi NC
- Subjects
- Cell Line, Tumor, DNA Helicases genetics, DNA-Binding Proteins genetics, Humans, Transcription, Genetic genetics, Xeroderma Pigmentosum genetics, DNA Repair genetics, Multiple Myeloma genetics
- Abstract
Despite the development of novel drugs, alkylating agents remain an important component of therapy in multiple myeloma (MM). DNA repair processes contribute towards sensitivity to alkylating agents and therefore we here evaluate the role of nucleotide excision repair (NER), which is involved in the removal of bulky adducts and DNA crosslinks in MM. We first evaluated NER activity using a novel functional assay and observed a heterogeneous NER efficiency in MM cell lines and patient samples. Using next-generation sequencing data, we identified that expression of the canonical NER gene, excision repair cross-complementation group 3 (ERCC3), significantly impacted the outcome in newly diagnosed MM patients treated with alkylating agents. Next, using small RNA interference, stable knockdown and overexpression, and small-molecule inhibitors targeting xeroderma pigmentosum complementation group B (XPB), the DNA helicase encoded by ERCC3, we demonstrate that NER inhibition significantly increases sensitivity and overcomes resistance to alkylating agents in MM. Moreover, inhibiting XPB leads to the dual inhibition of NER and transcription and is particularly efficient in myeloma cells. Altogether, we show that NER impacts alkylating agents sensitivity in myeloma cells and identify ERCC3 as a potential therapeutic target in MM.
- Published
- 2018
- Full Text
- View/download PDF
27. Risk Assessment to Evaluate if Crayons Complying with the Consumer Product Safety Improvement Act of 2008 for Lead, Also Comply with California Proposition 65.
- Author
-
Ahmad GR, Kumar S, Ahmad D, and Shammas MA
- Published
- 2017
- Full Text
- View/download PDF
28. Redefining Board Certified Toxicologist by Consumer Products Safety Commission May Increase Potential Risk of Exposure to Carcinogens among Consumers in United States of America.
- Author
-
Unmack JL, Bud Jenkins VC, and Shammas MA
- Published
- 2017
- Full Text
- View/download PDF
29. Impact of RAD51C-mediated Homologous Recombination on Genomic Integrity in Barrett's Adenocarcinoma Cells.
- Author
-
Pal J, Nanjappa P, Kumar S, Shi J, Buon L, Munshi NC, and Shammas MA
- Abstract
Background: In normal cells, RAD51-mediated homologous recombination (HR) is a precise DNA repair mechanism which plays a key role in the maintenance of genomic integrity and stability. However, elevated (dysregulated) RAD51 is implicated in genomic instability and is a potential target for treatment of certain cancers, including Barrett's adenocarcinoma (BAC). In this study, we investigated genomic impact and translational significance of moderate vs. strong suppression of RAD51 in BAC cells., Methods: BAC cells (FLO-1 and OE33) were transduced with non-targeting control (CS) or RAD51-specific shRNAs, mediating a moderate (40-50%) suppression or strong (80-near 100%) suppression of the gene. DNA breaks, spontaneous or following exposure to DNA damaging agent, were examined by comet assay and 53BP1 staining. Gene expression was monitored by microarrays (Affymetrix). Homologous recombination (HR) and single strand annealing (SSA) activities were measured using plasmid based assays., Results: We show that although moderate suppression consistenly inhibits/reduces HR activity, the strong suppression is associated with increase in HR activity (by ~15 - ≥ 50% in various experiments), suggesting activation of RAD51-independent pathway. Contrary to moderate suppression, a strong suppression of RAD51 is associated with a significant induced DNA breaks as well as altered expression of genes involved in detection/processing of DNA breaks and apoptosis. Stronger RAD51 suppression was also associated with mutagenic single strand annealing mediated HR. Suppression of RAD51C inhibited RAD51-independent (SSA-mediated) HR in BAC cells., Conclusion: Elevated (dysregulated) RAD51 in BAC is implicated in both the repair of DNA breaks as well as ongoing genomic rearrangements. Moderate suppression of this gene reduces HR activity, whereas strong or near complete suppression of this gene activates RAD51C-dependent HR involving a mechanism known as single strand annealing (SSA). SSA-mediated HR, which is a mutagenic HR pathway, further disrupts genomic integrity by increasing DNA breaks in BAC cells., Competing Interests: Conflict-of-interest statement: The author(s) declare(s) that there is no conflict of interest regarding the publication of this paper.
- Published
- 2017
- Full Text
- View/download PDF
30. Inclusion of "Toxicological Review Expiry Dates" in Art Material Labels May Further Reduce the Risk of Chronic Toxicity, Including That of Cancer.
- Author
-
Shammas MA, Rajput SA, Ahmad D, Ahmed M, Mustafa Z, and Ahmad G
- Published
- 2016
- Full Text
- View/download PDF
31. High frequency and poor prognosis of late childhood BCR-ABL-positive and MLL-AF4-positive ALL define the need for advanced molecular diagnostics and improved therapeutic strategies in pediatric B-ALL in Pakistan.
- Author
-
Iqbal Z, Akhtar T, Awan T, Aleem A, Sabir N, Rasool M, Absar M, Akram AM, Shammas MA, Shah IH, Khalid M, Taj AS, Jameel A, Alanazi A, Gill AT, Hashmi JA, Hussain A, Sabar MF, Khalid AM, Qazi MH, Karim S, Siddiqi MH, Mahmood A, Iqbal M, Saeed A, and Irfan MI
- Subjects
- Adolescent, Child, Child, Preschool, Female, Humans, In Situ Hybridization, Fluorescence, Infant, Male, Pakistan ethnology, Precision Medicine, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ethnology, Prognosis, Survival Analysis, Treatment Outcome, Fusion Proteins, bcr-abl genetics, Myeloid-Lymphoid Leukemia Protein genetics, Oncogene Proteins, Fusion genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
Background: Fusion oncogenes (FOs) resulting from chromosomal abnormalities have an important role in leukemogenesis in pediatric B cell acute lymphoblastic leukemia (ALL). The most common FOs are BCR-ABL, MLL-AF4, ETV6-RUNX1, and TCF3-PBX1, all of which have important prognostic and drug selection implications. Moreover, frequencies of FOs have ethnic variations. We studied Pakistani frequencies of FOs, clinical pattern, and outcome in pediatric B-ALL., Methods: FOs were studied in 188 patients at diagnosis using reverse transcriptase-polymerase chain reaction (RT-PCR) and interphase fluorescent in situ hybridization (FISH). Data were analyzed using SPSS version 17 (SPSS Inc., Chicago, IL, USA)., Results: FOs were detected in 87.2 % of patients. Mean overall survival was 70.9 weeks, 3-year survival was 31.9 %, and 3-year relapse-free survival was 18.1 %. Four patients died of drug toxicities. ETV6-RUNX1 (19.14 %) had better survival (110.9 weeks; p = 0.03); TCF3-PBX1 (2.1 %) was associated with inferior outcome and higher central nervous system (CNS) relapse risk; MLL-AF4 (18.1 %) was more common in the 8- to 15-year age group (24/34; p = 0.001) and was associated with organomegaly, low platelet count, and poor survival; and BCR-ABL (47.9 %) was associated with older age (7-15 years, 52/90), lower remission rates, shorter survival (43.73 ± 4.24 weeks) and higher white blood cell count. Overall, MLL-AF4 and BCR-ABL were detected in 66 % of B-ALL, presented in later childhood, and were associated with poor prognosis and inferior survival., Conclusions: This study reports the highest ethnic frequency of BCR-ABL FO in pediatric ALL, and is consistent with previous reports from our region. Poor prognosis BCR-ABL and MLL-AF4 was detected in two-thirds of pediatric B-ALL and is likely to be the reason for the already reported poor survival of childhood ALL in South-East Asia. Furthermore, MLL-AF4, usually most common in infants, presented in later childhood in most of the ALL patients, which was one of the unique findings in our study. The results presented here highlight the need for mandatory inclusion of molecular testing for pediatric ALL patients in clinical decision making, together with the incorporation of tyrosine kinase inhibitors, as well as hematopoietic stem cell transplantation facilities, to improve treatment outcome for patients in developing countries.
- Published
- 2015
- Full Text
- View/download PDF
32. Suggested safe harbor limit for titanium dioxide: an exposure level which protects consumers from cancer incidence.
- Author
-
Shammas MA, Ahmad D, Nguyen MD, Rajput S, Unmack J, and Ahmad G
- Published
- 2015
- Full Text
- View/download PDF
33. Differential and limited expression of mutant alleles in multiple myeloma.
- Author
-
Rashid NU, Sperling AS, Bolli N, Wedge DC, Van Loo P, Tai YT, Shammas MA, Fulciniti M, Samur MK, Richardson PG, Magrangeas F, Minvielle S, Futreal PA, Anderson KC, Avet-Loiseau H, Campbell PJ, Parmigiani G, and Munshi NC
- Subjects
- Alleles, DNA genetics, Humans, RNA genetics, Sequence Analysis, RNA, Gene Expression Regulation, Neoplastic, Multiple Myeloma genetics, Mutation
- Abstract
Recent work has delineated mutational profiles in multiple myeloma and reported a median of 52 mutations per patient, as well as a set of commonly mutated genes across multiple patients. In this study, we have used deep sequencing of RNA from a subset of these patients to evaluate the proportion of expressed mutations. We find that the majority of previously identified mutations occur within genes with very low or no detectable expression. On average, 27% (range, 11% to 47%) of mutated alleles are found to be expressed, and among mutated genes that are expressed, there often is allele-specific expression where either the mutant or wild-type allele is suppressed. Even in the absence of an overall change in gene expression, the presence of differential allelic expression within malignant cells highlights the important contribution of RNA-sequencing in identifying clinically significant mutational changes relevant to our understanding of myeloma biology and also for therapeutic applications.
- Published
- 2014
- Full Text
- View/download PDF
34. Targeting homologous recombination and telomerase in Barrett's adenocarcinoma: impact on telomere maintenance, genomic instability and tumor growth.
- Author
-
Lu R, Pal J, Buon L, Nanjappa P, Shi J, Fulciniti M, Tai YT, Guo L, Yu M, Gryaznov S, Munshi NC, and Shammas MA
- Subjects
- Adenocarcinoma complications, Adenocarcinoma drug therapy, Adenocarcinoma pathology, Animals, Antineoplastic Combined Chemotherapy Protocols, Barrett Esophagus enzymology, Barrett Esophagus genetics, Barrett Esophagus pathology, Cell Line, Tumor, Cell Proliferation drug effects, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Esophageal Neoplasms complications, Esophageal Neoplasms drug therapy, Gene Knockout Techniques, Humans, Male, Mice, Oligonucleotides metabolism, Pyrimidines pharmacology, Pyrimidines therapeutic use, Rad51 Recombinase deficiency, Rad51 Recombinase genetics, Telomerase metabolism, Telomere genetics, Adenocarcinoma genetics, Barrett Esophagus complications, Esophageal Neoplasms genetics, Esophageal Neoplasms pathology, Genomic Instability drug effects, Homologous Recombination drug effects, Telomerase antagonists & inhibitors, Telomere drug effects
- Abstract
Homologous recombination (HR), a mechanism to accurately repair DNA in normal cells, is deregulated in cancer. Elevated/deregulated HR is implicated in genomic instability and telomere maintenance, which are critical lifelines of cancer cells. We have previously shown that HR activity is elevated and significantly contributes to genomic instability in Barrett's esophageal adenocarcinoma (BAC). The purpose of this study was to evaluate therapeutic potential of HR inhibition, alone and in combination with telomerase inhibition, in BAC. We demonstrate that telomerase inhibition in BAC cells increases HR activity, RAD51 expression, and association of RAD51 to telomeres. Suppression of HR leads to shorter telomeres as well as markedly reduced genomic instability in BAC cells over time. Combination of HR suppression (whether transgenic or chemical) with telomerase inhibition, causes a significant increase in telomere attrition and apoptotic death in all BAC cell lines tested, relative to either treatment alone. A subset of treated cells also stain positive for β-galactosidase, indicating senescence. The combined treatment is also associated with decline in S-phase and a strong G2/M arrest, indicating massive telomere attrition. In a subcutaneous tumor model, the combined treatment resulted in the smallest tumors, which were even smaller (P=0.001) than those that resulted from either treatment alone. Even the tumors removed from these mice had significantly reduced telomeres and evidence of apoptosis. We therefore conclude that although telomeres are elongated by telomerase, elevated RAD51/HR assist in their maintenance/stabilization in BAC cells. Telomerase inhibitor prevents telomere elongation but induces RAD51/HR, which contributes to telomere maintenance/stabilization and prevention of apoptosis, reducing the efficacy of treatment. Combining HR inhibition with telomerase renders telomeres more vulnerable to degradation and significantly increases/expedites their attrition, leading to apoptosis. We therefore demonstrate that a therapy targeting HR and telomerase has the potential to prevent both tumor growth and genomic evolution in BAC.
- Published
- 2014
- Full Text
- View/download PDF
35. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma.
- Author
-
Bolli N, Avet-Loiseau H, Wedge DC, Van Loo P, Alexandrov LB, Martincorena I, Dawson KJ, Iorio F, Nik-Zainal S, Bignell GR, Hinton JW, Li Y, Tubio JM, McLaren S, O' Meara S, Butler AP, Teague JW, Mudie L, Anderson E, Rashid N, Tai YT, Shammas MA, Sperling AS, Fulciniti M, Richardson PG, Parmigiani G, Magrangeas F, Minvielle S, Moreau P, Attal M, Facon T, Futreal PA, Anderson KC, Campbell PJ, and Munshi NC
- Subjects
- Adult, Aged, Antigens, Nuclear, Cohort Studies, DNA Copy Number Variations, Early Growth Response Protein 1, Evolution, Molecular, GTP Phosphohydrolases, Genetic Heterogeneity, Humans, Lymphotoxin-beta, Membrane Proteins, Middle Aged, Mutation, Mutation, Missense, Nerve Tissue Proteins, Proto-Oncogene Proteins, Proto-Oncogene Proteins B-raf, Proto-Oncogene Proteins p21(ras), Receptors, Immunologic, Sequence Analysis, DNA, Transcription Factors, Tumor Suppressor Protein p53, ras Proteins, Roundabout Proteins, Exome, Multiple Myeloma genetics
- Abstract
Multiple myeloma is an incurable plasma cell malignancy with a complex and incompletely understood molecular pathogenesis. Here we use whole-exome sequencing, copy-number profiling and cytogenetics to analyse 84 myeloma samples. Most cases have a complex subclonal structure and show clusters of subclonal variants, including subclonal driver mutations. Serial sampling reveals diverse patterns of clonal evolution, including linear evolution, differential clonal response and branching evolution. Diverse processes contribute to the mutational repertoire, including kataegis and somatic hypermutation, and their relative contribution changes over time. We find heterogeneity of mutational spectrum across samples, with few recurrent genes. We identify new candidate genes, including truncations of SP140, LTB, ROBO1 and clustered missense mutations in EGR1. The myeloma genome is heterogeneous across the cohort, and exhibits diversity in clonal admixture and in dynamics of evolution, which may impact prognostic stratification, therapeutic approaches and assessment of disease response to treatment.
- Published
- 2014
- Full Text
- View/download PDF
36. Biology of telomeres: importance in etiology of esophageal cancer and as therapeutic target.
- Author
-
Pal J, Gold JS, Munshi NC, and Shammas MA
- Subjects
- Adenocarcinoma etiology, Adenocarcinoma therapy, Esophageal Neoplasms therapy, Genomic Instability, Homologous Recombination, Humans, Risk Factors, Telomerase genetics, Telomerase physiology, Telomere genetics, Telomere Shortening, Translational Research, Biomedical, Esophageal Neoplasms etiology, Telomere physiology
- Abstract
The purpose of this review is to highlight the importance of telomeres, the mechanisms implicated in their maintenance, and their role in the etiology as well as the treatment of human esophageal cancer. We will also discuss the role of telomeres in the maintenance and preservation of genomic integrity, the consequences of telomere dysfunction, and the various factors that may affect telomere health in esophageal tissue predisposing it to oncogenesis. There has been growing evidence that telomeres, which can be affected by various intrinsic and extrinsic factors, contribute to genomic instability, oncogenesis, as well as proliferation of cancer cells. Telomeres are the protective DNA-protein complexes at chromosome ends. Telomeric DNA undergoes progressive shortening with age leading to cellular senescence and/or apoptosis. If senescence/apoptosis is prevented as a consequence of specific genomic changes, continued proliferation leads to very short (ie, dysfunctional) telomeres that can potentially cause genomic instability, thus, increasing the risk for activation of telomere maintenance mechanisms and oncogenesis. Like many other cancers, esophageal cancer cells have short telomeres and elevated telomerase, the enzyme that maintains telomeres in most cancer cells. Homologous recombination, which is implicated in the alternate pathway of telomere elongation, is also elevated in Barrett's-associated esophageal adenocarcinoma. Evidence from our laboratory indicates that both telomerase and homologous recombination contribute to telomere maintenance, DNA repair, and the ongoing survival of esophageal cancer cells. This indicates that telomere maintenance mechanisms may potentially be targeted to make esophageal cancer cells static. The rate at which telomeres in healthy cells shorten is determined by a number of intrinsic and extrinsic factors, including those associated with lifestyle. Avoidance of factors that may directly or indirectly injure esophageal tissue including its telomeric and other genomic DNA can not only reduce the risk of development of esophageal cancer but may also have positive impact on overall health and lifespan., (Copyright © 2013 Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. Synthetic miR-34a mimics as a novel therapeutic agent for multiple myeloma: in vitro and in vivo evidence.
- Author
-
Di Martino MT, Leone E, Amodio N, Foresta U, Lionetti M, Pitari MR, Cantafio ME, Gullà A, Conforti F, Morelli E, Tomaino V, Rossi M, Negrini M, Ferrarini M, Caraglia M, Shammas MA, Munshi NC, Anderson KC, Neri A, Tagliaferri P, and Tassone P
- Subjects
- Animals, Apoptosis, Cell Line, Cell Proliferation, Genes, Tumor Suppressor, Genetic Therapy, Humans, Lentivirus genetics, Male, Mice, Mice, SCID, MicroRNAs biosynthesis, Multiple Myeloma genetics, Multiple Myeloma pathology, Neoplasm Transplantation, RNA Interference, Transduction, Genetic, Transfection, Tumor Burden, Tumor Microenvironment, MicroRNAs genetics, Multiple Myeloma therapy
- Abstract
Purpose: Deregulated expression of miRNAs has been shown in multiple myeloma (MM). A promising strategy to achieve a therapeutic effect by targeting the miRNA regulatory network is to enforce the expression of miRNAs that act as tumor suppressor genes, such as miR-34a., Experimental Design: Here, we investigated the therapeutic potential of synthetic miR-34a against human MM cells in vitro and in vivo., Results: Either transient expression of miR-34a synthetic mimics or lentivirus-based miR-34a-stable enforced expression triggered growth inhibition and apoptosis in MM cells in vitro. Synthetic miR-34a downregulated canonic targets BCL2, CDK6, and NOTCH1 at both the mRNA and protein level. Lentiviral vector-transduced MM xenografts with constitutive miR-34a expression showed high growth inhibition in severe combined immunodeficient (SCID) mice. The anti-MM activity of lipidic-formulated miR-34a was further shown in vivo in two different experimental settings: (i) SCID mice bearing nontransduced MM xenografts; and (ii) SCID-synth-hu mice implanted with synthetic 3-dimensional scaffolds reconstituted with human bone marrow stromal cells and then engrafted with human MM cells. Relevant tumor growth inhibition and survival improvement were observed in mice bearing TP53-mutated MM xenografts treated with miR-34a mimics in the absence of systemic toxicity., Conclusions: Our findings provide a proof-of-principle that formulated synthetic miR-34a has therapeutic activity in preclinical models and support a framework for development of miR-34a-based treatment strategies in MM patients., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
38. Targeting PI3K and RAD51 in Barrett's adenocarcinoma: impact on DNA damage checkpoints, expression profile and tumor growth.
- Author
-
Pal J, Fulciniti M, Nanjappa P, Buon L, Tai YT, Tassone P, Munshi NC, and Shammas MA
- Subjects
- Adenocarcinoma enzymology, Androstadienes pharmacology, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cluster Analysis, Esophageal Neoplasms enzymology, Gene Expression drug effects, Male, Mice, Mice, SCID, Phosphatidylinositol 3-Kinase genetics, Phosphatidylinositol 3-Kinase metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering metabolism, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Wortmannin, Adenocarcinoma genetics, DNA Damage, Esophageal Neoplasms genetics, Gene Expression Profiling, Phosphoinositide-3 Kinase Inhibitors, Rad51 Recombinase antagonists & inhibitors
- Abstract
Phosphatidylinositol 3-kinase (PI3K)/v-akt murine thymoma viral oncogene homolog 1 (AKT) signaling in cancer is implicated in various survival pathways including regulation of recombinase (RAD51). In this study, we evaluated PI3K and RAD51 as targets in Barrett's adenocarcinoma (BAC) cells both in vitro and in vivo. BAC cell lines (OE19, OE33, and FLO-1) were cultured in the presence of PI3K inhibitor (wortmannin) and the impact on growth and expression of AKT, phosphorylated-AKT (P-AKT), and RAD51 was determined. Wortmannin induced growth arrest and apoptosis in two BAC cell lines (OE33 and OE19), which had relatively higher expression of AKT. FLO-1 cells, with lower AKT expression, were less sensitive to treatment and investigated further. In FLO-1 cells, wortmannin suppressed ataxia telangiectasia and Rad3-related protein (ATR)-checkpoint kinase 1 (CHK1)-mediated checkpoint and multiple DNA repair genes, whereas RAD51 and CHK2 were not affected. Western blotting confirmed that RAD51 was suppressed by wortmannin in OE33 and OE19 cells, but not in FLO-1 cells. Suppression of RAD51 in FLO-1 cells down-regulated the expression of CHK2 and CHK1, and reduced the proliferative potential. Finally, the suppression of RAD51 in FLO-1 cells, significantly increased the anticancer activity of wortmannin in these cells, both in vitro and in vivo. We show that PI3K signaling and hsRAD51, through distinct roles in DNA damage response and repair pathways, provide survival advantage to BAC cells. In cells with inherent low expression of AKT, RAD51 is unaffected by PI3K suppression and provides an additional survival pathway. Simultaneous suppression of PI3K and RAD51, especially in cells with lower AKT expression, can significantly reduce their proliferative potential.
- Published
- 2012
39. Repetitive sequences, genomic instability and Barrett's esophageal adenocarcinoma.
- Author
-
Shammas MA
- Abstract
Barrett's esophageal adenocarcinoma (BAC) is a cancer associated with heartburn. If gastroesophageal reflux is not treated, the exposure to acid over the years, leads to a premalignant condition known as Barrett's esophagus (BE) which then progresses through low grade and high grade dysplasias to Barrett's adenocarcinoma. Genomic instability, which seems to arise early at BE stage, leads to accrual of mutational changes which underlie the the succession of histological and physiological changes associated with this disease. Genomic instability is therefore an important target for prevention and treatment of cancer and it is important to elucidate the mechanisms associated with this problem. We have shown that elevated/deregulated homologous recombination mediates genomic instability in cancer. Recently we also demonstrated that the mutational rates of individual chromosomes in BAC cells correlate with their ALU frequency. The aims of this article are to briefly discuss different types of repetitive sequences and highlight their importance in physiology of normal and cancer cells, especially BAC.
- Published
- 2011
- Full Text
- View/download PDF
40. Purification of diseased cells from Barrett's esophagus and related lesions by laser capture microdissection.
- Author
-
Shammas MA and Rao MY
- Subjects
- Barrett Esophagus metabolism, Cryopreservation, DNA isolation & purification, Enzyme Assays, Epithelial Cells pathology, Esophagus pathology, Humans, Paraffin Embedding, RNA isolation & purification, Telomerase chemistry, Telomerase metabolism, Barrett Esophagus pathology, Cell Separation methods, Lasers, Microdissection methods
- Abstract
Barrett's esophageal adenocarcinoma (BEAC) arises from Barrett's esophagus (BE), a premalignant lesion caused by acid reflux (heartburn). Although the cancer is uncommon, its incidence is rapidly rising in western countries. Like most other cancers, BEAC cells also have elevated telomerase activity which maintains telomere length and supports continued proliferation of these cells. It is not clear if telomerase is activated early at premalignant (BE) stage, because reports of telomerase activity in Barrett's and normal esophagi have been controversial. We have shown that detection of telomerase and telomeres becomes easier and much more reliable if purified BE cells are used instead of tissue specimens. This chapter, therefore, emphasizes the importance of laser capture microdissection and provides the method to purify Barrett's esophagus related cells, using this technique.
- Published
- 2011
- Full Text
- View/download PDF
41. Anticancer activity of a broccoli derivative, sulforaphane, in barrett adenocarcinoma: potential use in chemoprevention and as adjuvant in chemotherapy.
- Author
-
Qazi A, Pal J, Maitah M, Fulciniti M, Pelluru D, Nanjappa P, Lee S, Batchu RB, Prasad M, Bryant CS, Rajput S, Gryaznov S, Beer DG, Weaver DW, Munshi NC, Goyal RK, and Shammas MA
- Abstract
Introduction: The incidence of Barrett esophageal adenocarcinoma (BEAC) has been increasing at an alarming rate in western countries. In this study, we have evaluated the therapeutic potential of sulforaphane (SFN), an antioxidant derived from broccoli, in BEAC., Methods: BEAC cells were treated with SFN, alone or in combination with chemotherapeutic, paclitaxel, or telomerase-inhibiting agents (MST-312, GRN163L), and live cell number determined at various time points. The effect on drug resistance/chemosensitivity was evaluated by rhodamine efflux assay. Apoptosis was detected by annexin V labeling and Western blot analysis of poly(ADP-ribose) polymerase cleavage. Effects on genes implicated in cell cycle and apoptosis were determined by Western blot analyses. To evaluate the efficacy in vivo, BEAC cells were injected subcutaneously in severe combined immunodeficient mice, and after the appearance of palpable tumors, mice were treated with SFN., Results: SFN induced both time- and dose-dependent decline in cell survival, cell cycle arrest, and apoptosis. The treatment with SFN also suppressed the expression of multidrug resistance protein, reduced drug efflux, and increased anticancer activity of other antiproliferative agents including paclitaxel. A significant reduction in tumor volume was also observed by SFN in a subcutaneous tumor model of BEAC. Anticancer activity could be attributed to the induction of caspase 8 and p21 and down-regulation of hsp90, a molecular chaperon required for activity of several proliferation-associated proteins., Conclusions: These data indicate that a natural product with antioxidant properties from broccoli has great potential to be used in chemoprevention and treatment of BEAC.
- Published
- 2010
- Full Text
- View/download PDF
42. Sulforaphane induces cell cycle arrest by protecting RB-E2F-1 complex in epithelial ovarian cancer cells.
- Author
-
Bryant CS, Kumar S, Chamala S, Shah J, Pal J, Haider M, Seward S, Qazi AM, Morris R, Semaan A, Shammas MA, Steffes C, Potti RB, Prasad M, Weaver DW, and Batchu RB
- Subjects
- Apoptosis drug effects, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, DNA, Neoplasm biosynthesis, Epithelial Cells drug effects, Epithelial Cells metabolism, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Isothiocyanates, Neoplasm Invasiveness, Oligonucleotide Array Sequence Analysis, Ovarian Neoplasms genetics, Paclitaxel pharmacology, Phosphorylation drug effects, S Phase drug effects, Sulfoxides, Wound Healing drug effects, Cell Cycle drug effects, E2F1 Transcription Factor metabolism, Epithelial Cells pathology, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Retinoblastoma Protein metabolism, Thiocyanates pharmacology
- Abstract
Background: Sulforaphane (SFN), an isothiocyanate phytochemical present predominantly in cruciferous vegetables such as brussels sprout and broccoli, is considered a promising chemo-preventive agent against cancer. In-vitro exposure to SFN appears to result in the induction of apoptosis and cell-cycle arrest in a variety of tumor types. However, the molecular mechanisms leading to the inhibition of cell cycle progression by SFN are poorly understood in epithelial ovarian cancer cells (EOC). The aim of this study is to understand the signaling mechanisms through which SFN influences the cell growth and proliferation in EOC., Results: SFN at concentrations of 5-20 microM induced a dose-dependent suppression of growth in cell lines MDAH 2774 and SkOV-3 with an IC50 of ~8 microM after a 3 day exposure. Combination treatment with chemotherapeutic agent, paclitaxel, resulted in additive growth suppression. SFN at ~8 microM decreased growth by 40% and 20% on day 1 in MDAH 2774 and SkOV-3, respectively. Cells treated with cytotoxic concentrations of SFN have reduced cell migration and increased apoptotic cell death via an increase in Bak/Bcl-2 ratio and cleavage of procaspase-9 and poly (ADP-ribose)-polymerase (PARP). Gene expression profile analysis of cell cycle regulated proteins demonstrated increased levels of tumor suppressor retinoblastoma protein (RB) and decreased levels of E2F-1 transcription factor. SFN treatment resulted in G1 cell cycle arrest through down modulation of RB phosphorylation and by protecting the RB-E2F-1 complex., Conclusions: SFN induces growth arrest and apoptosis in EOC cells. Inhibition of retinoblastoma (RB) phosphorylation and reduction in levels of free E2F-1 appear to play an important role in EOC growth arrest.
- Published
- 2010
- Full Text
- View/download PDF
43. Ritonavir blocks AKT signaling, activates apoptosis and inhibits migration and invasion in ovarian cancer cells.
- Author
-
Kumar S, Bryant CS, Chamala S, Qazi A, Seward S, Pal J, Steffes CP, Weaver DW, Morris R, Malone JM, Shammas MA, Prasad M, and Batchu RB
- Subjects
- Biomarkers, Tumor metabolism, Blotting, Western, Caspase 3 metabolism, Cell Proliferation drug effects, Cyclin-Dependent Kinases metabolism, Cyclins metabolism, Female, Gene Expression Profiling, Humans, Neoplasm Invasiveness, Oligonucleotide Array Sequence Analysis, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Phosphorylation drug effects, Poly(ADP-ribose) Polymerases metabolism, Proto-Oncogene Proteins c-akt genetics, RNA, Small Interfering pharmacology, Retinoblastoma Protein metabolism, Signal Transduction drug effects, Tumor Cells, Cultured, Wound Healing drug effects, Apoptosis drug effects, Cell Cycle drug effects, Cell Movement drug effects, HIV Protease Inhibitors pharmacology, Ovarian Neoplasms pathology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Ritonavir pharmacology
- Abstract
Background: Ovarian cancer is the leading cause of mortality from gynecological malignancies, often undetectable in early stages. The difficulty of detecting the disease in its early stages and the propensity of ovarian cancer cells to develop resistance to known chemotherapeutic treatments dramatically decreases the 5-year survival rate. Chemotherapy with paclitaxel after surgery increases median survival only by 2 to 3 years in stage IV disease highlights the need for more effective drugs. The human immunodeficiency virus (HIV) infection is characterized by increased risk of several solid tumors due to its inherent nature of weakening of immune system. Recent observations point to a lower incidence of some cancers in patients treated with protease inhibitor (PI) cocktail treatment known as HAART (Highly Active Anti-Retroviral Therapy)., Results: Here we show that ritonavir, a HIV protease inhibitor effectively induced cell cycle arrest and apoptosis in ovarian cell lines MDH-2774 and SKOV-3 in a dose dependent manner. Over a 3 day period with 20 muM ritonavir resulted in the cell death of over 60% for MDAH-2774 compared with 55% in case of SKOV-3 cell line. Ritonavir caused G1 cell cycle arrest of the ovarian cancer cells, mediated by down modulating levels of RB phosphorylation and depleting the G1 cyclins, cyclin-dependent kinase and increasing their inhibitors as determined by gene profile analysis. Interestingly, the treatment of ritonavir decreased the amount of phosphorylated AKT in a dose-dependent manner. Furthermore, inhibition of AKT by specific siRNA synergistically increased the efficacy of the ritonavir-induced apoptosis. These results indicate that the addition of the AKT inhibitor may increase the therapeutic efficacy of ritonavir., Conclusion: Our results demonstrate a potential use of ritonavir for ovarian cancer with additive effects in conjunction with conventional chemotherapeutic regimens. Since ritonavir is clinically approved for human use for HIV, drug repositioning for ovarian cancer could accelerate the process of traditional drug development. This would reduce risks, limit the costs and decrease the time needed to bring the drug from bench to bedside.
- Published
- 2009
- Full Text
- View/download PDF
44. Dysfunctional homologous recombination mediates genomic instability and progression in myeloma.
- Author
-
Shammas MA, Shmookler Reis RJ, Koley H, Batchu RB, Li C, and Munshi NC
- Subjects
- DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Disease Progression, Humans, Immunohistochemistry, Loss of Heterozygosity, Mutation, Polymerase Chain Reaction, Promoter Regions, Genetic, RNA, Small Interfering, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Transfection, Gene Expression Profiling, Genomic Instability, Multiple Myeloma genetics, Recombination, Genetic
- Abstract
A prominent feature of most if not all cancers is a striking genetic instability, leading to ongoing accrual of mutational changes, some of which underlie tumor progression, including acquisition of invasiveness, drug resistance, and metastasis. Thus, the molecular basis for the generation of this genetic diversity in cancer cells has important implications in understanding cancer progression. Here we report that homologous recombination (HR) activity is elevated in multiple myeloma (MM) cells and leads to an increased rate of mutation and progressive accumulation of genetic variation over time. We demonstrate that the inhibition of HR activity in MM cells by small inhibitory RNA (siRNAs) targeting recombinase leads to significant reduction in the acquisition of new genetic changes in the genome and, conversely, the induction of HR activity leads to significant elevation in the number of new mutations over time and development of drug resistance in MM cells. These data identify dysregulated HR activity as a key mediator of DNA instability and progression of MM, with potential as a therapeutic target.
- Published
- 2009
- Full Text
- View/download PDF
45. Generation of antitumor invariant natural killer T cell lines in multiple myeloma and promotion of their functions via lenalidomide: a strategy for immunotherapy.
- Author
-
Song W, van der Vliet HJ, Tai YT, Prabhala R, Wang R, Podar K, Catley L, Shammas MA, Anderson KC, Balk SP, Exley MA, and Munshi NC
- Subjects
- Antigens, CD1 immunology, Cytotoxicity, Immunologic, Humans, Immunotherapy methods, Lenalidomide, Thalidomide pharmacology, Cell Line, Killer Cells, Natural, Multiple Myeloma immunology, Thalidomide analogs & derivatives
- Abstract
Purpose: CD1d-restricted invariant natural killer T (iNKT) cells are important immunoregulatory cells in antitumor immune responses. However, the quantitative and qualitative defects of iNKT cells in advanced multiple myeloma hamper their antitumor effects. Therefore, the development of functional iNKT cells may provide a novel strategy for the immunotherapy in multiple myeloma., Experimental Design: We activated and expanded iNKT cells from multiple myeloma patients with alpha-galactosylceramide (alpha-GalCer)-pulsed dendritic cells, characterized their antitumor effects by the cytokine production profile and cytotoxicity against multiple myeloma cells, and explored the effects of immunomodulatory drug lenalidomide on these iNKT cells. We also investigated the expression of CD1d by primary multiple myeloma cells and its function to activate iNKT cells., Results: We established highly purified functional iNKT cell lines from newly diagnosed and advanced multiple myeloma patients. These CD1d-restricted iNKT cell lines produced high level of antitumor Th1 cytokine in response to alpha-GalCer-pulsed primary multiple myeloma cells, CD1d-transfected MM1S cell line, and dendritic cells. Moreover, iNKT cell lines displayed strong cytotoxicity against alpha-GalCer-pulsed primary multiple myeloma cells. Importantly, lenalidomide further augmented the Th1 polarization by iNKT cell lines via increased Th1 cytokine production and reduced Th2 cytokine production. We also showed that CD1d was expressed in primary multiple myeloma cells at mRNA and protein levels from the majority of multiple myeloma patients, but not in normal plasma cells and multiple myeloma cell lines, and CD1d(+) primary multiple myeloma cells presented antigens to activate iNKT cell lines., Conclusions: Taken together, our results provide the preclinical evidence for the iNKT cell-mediated immunotherapy and a rationale for their use in combination with lenalidomide in multiple myeloma treatment.
- Published
- 2008
- Full Text
- View/download PDF
46. Telomere maintenance in laser capture microdissection-purified Barrett's adenocarcinoma cells and effect of telomerase inhibition in vivo.
- Author
-
Shammas MA, Qazi A, Batchu RB, Bertheau RC, Wong JY, Rao MY, Prasad M, Chanda D, Ponnazhagan S, Anderson KC, Steffes CP, Munshi NC, De Vivo I, Beer DG, Gryaznov S, Weaver DW, and Goyal RK
- Subjects
- Adenocarcinoma metabolism, Animals, Apoptosis, Barrett Esophagus metabolism, Cell Line, Tumor, Cellular Senescence, Epithelial Cells ultrastructure, Female, Humans, Lasers, Mice, Mice, SCID, Microdissection, Neoplasm Transplantation, Telomerase metabolism, Adenocarcinoma ultrastructure, Barrett Esophagus ultrastructure, Telomerase antagonists & inhibitors, Telomere ultrastructure
- Abstract
Purpose: The aims of this study were to investigate telomere function in normal and Barrett's esophageal adenocarcinoma (BEAC) cells purified by laser capture microdissection and to evaluate the effect of telomerase inhibition in cancer cells in vitro and in vivo., Experimental Design: Epithelial cells were purified from surgically resected esophagi. Telomerase activity was measured by modified telomeric repeat amplification protocol and telomere length was determined by real-time PCR assay. To evaluate the effect of telomerase inhibition, adenocarcinoma cell lines were continuously treated with a specific telomerase inhibitor (GRN163L) and live cell number was determined weekly. Apoptosis was evaluated by Annexin labeling and senescence by beta-galactosidase staining. For in vivo studies, severe combined immunodeficient mice were s.c. inoculated with adenocarcinoma cells and following appearance of palpable tumors, injected i.p. with saline or GRN163L., Results: Telomerase activity was significantly elevated whereas telomeres were shorter in BEAC cells relative to normal esophageal epithelial cells. The treatment of adenocarcinoma cells with telomerase inhibitor, GRN163L, led to loss of telomerase activity, reduction in telomere length, and growth arrest through induction of both the senescence and apoptosis. GRN163L-induced cell death could also be expedited by addition of the chemotherapeutic agents doxorubicin and ritonavir. Finally, the treatment with GRN163L led to a significant reduction in tumor volume in a subcutaneous tumor model., Conclusions: We show that telomerase activity is significantly elevated whereas telomeres are shorter in BEAC and suppression of telomerase inhibits proliferation of adenocarcinoma cells both in vitro and in vivo.
- Published
- 2008
- Full Text
- View/download PDF
47. Telomerase inhibitor GRN163L inhibits myeloma cell growth in vitro and in vivo.
- Author
-
Shammas MA, Koley H, Bertheau RC, Neri P, Fulciniti M, Tassone P, Blotta S, Protopopov A, Mitsiades C, Batchu RB, Anderson KC, Chin A, Gryaznov S, and Munshi NC
- Subjects
- Animals, Apoptosis drug effects, Benzoquinones pharmacology, Cell Line, Tumor, Gene Expression Profiling, HSP90 Heat-Shock Proteins antagonists & inhibitors, Humans, Lactams, Macrocyclic pharmacology, Mice, Mice, SCID, Multiple Myeloma genetics, Multiple Myeloma pathology, Oligonucleotides, Oligopeptides pharmacokinetics, Telomerase metabolism, Telomere, Enzyme Inhibitors pharmacology, Multiple Myeloma drug therapy, Oligopeptides pharmacology, Telomerase antagonists & inhibitors
- Abstract
Human telomerase, the reverse transcriptase which extends the life span of a cell by adding telomeric repeats to chromosome ends, is expressed in most cancer cells but not in the majority of normal somatic cells. Inhibition of telomerase therefore holds great promise as anticancer therapy. We have synthesized a novel telomerase inhibitor GRN163L, a lipid-attached phosphoramidate oligonucleotide complementary to template region of the RNA subunit of telomerase. Here, we report that GRN163L is efficiently taken up by human myeloma cells without any need of transfection and is resistant to nucleolytic degradation. The exposure of myeloma cells to GRN163L led to an effective inhibition of telomerase activity, reduction of telomere length and apoptotic cell death after a lag period of 2-3 weeks. Mismatch control oligonucleotides had no effect on growth of myeloma cells. The in vivo efficacy of GRN163L was confirmed in two murine models of human multiple myeloma. In three independent experiments, significant reduction in tumor cell growth and better survival than control mice was observed. Furthermore, GRN163L-induced myeloma cell death could be significantly enhanced by Hsp90 inhibitor 17AAG. These data provide the preclinical rationale for clinical evaluation of GRN163L in myeloma and in combination with 17AAG.
- Published
- 2008
- Full Text
- View/download PDF
48. Specific killing of multiple myeloma cells by (-)-epigallocatechin-3-gallate extracted from green tea: biologic activity and therapeutic implications.
- Author
-
Shammas MA, Neri P, Koley H, Batchu RB, Bertheau RC, Munshi V, Prabhala R, Fulciniti M, Tai YT, Treon SP, Goyal RK, Anderson KC, and Munshi NC
- Subjects
- Animals, Antineoplastic Agents isolation & purification, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Catechin isolation & purification, Catechin pharmacology, Catechin therapeutic use, Cell Line, Tumor, Gene Expression Profiling, Humans, Mice, Mice, SCID, Multiple Myeloma genetics, Multiple Myeloma metabolism, Multiple Myeloma pathology, Neoplasm Transplantation, Phytotherapy, RNA, Small Interfering genetics, Receptors, Laminin antagonists & inhibitors, Receptors, Laminin genetics, Receptors, Laminin metabolism, Tea chemistry, Transplantation, Heterologous, Catechin analogs & derivatives, Multiple Myeloma drug therapy
- Abstract
Epigallocatechin-3-gallate (EGCG), a polyphenol extracted from green tea, is an antioxidant with chemopreventive and chemotherapeutic actions. Based on its ability to modulate growth factor-mediated cell proliferation, we evaluated its efficacy in multiple myeloma (MM). EGCG induced both dose- and time-dependent growth arrest and subsequent apoptotic cell death in MM cell lines including IL-6-dependent cells and primary patient cells, without significant effect on the growth of peripheral blood mononuclear cells (PBMCs) and normal fibroblasts. Treatment with EGCG also led to significant apoptosis in human myeloma cells grown as tumors in SCID mice. EGCG interacts with the 67-kDa laminin receptor 1 (LR1), which is significantly elevated in myeloma cell lines and patient samples relative to normal PBMCs. RNAi-mediated inhibition of LR1 resulted in abrogation of EGCG-induced apoptosis in myeloma cells, indicating that LR1 plays an important role in mediating EGCG activity in MM while sparing PBMCs. Evaluation of changes in gene expression profile indicates that EGCG treatment activates distinct pathways of growth arrest and apoptosis in MM cells by inducing the expression of death-associated protein kinase 2, the initiators and mediators of death receptor-dependent apoptosis (Fas ligand, Fas, and caspase 4), p53-like proteins (p73, p63), positive regulators of apoptosis and NF-kappaB activation (CARD10, CARD14), and cyclin-dependent kinase inhibitors (p16 and p18). Expression of related genes at the protein level were also confirmed by Western blot analysis. These data demonstrate potent and specific antimyeloma activity of EGCG and provide the rationale for its clinical evaluation.
- Published
- 2006
- Full Text
- View/download PDF
49. Dysfunctional T regulatory cells in multiple myeloma.
- Author
-
Prabhala RH, Neri P, Bae JE, Tassone P, Shammas MA, Allam CK, Daley JF, Chauhan D, Blanchard E, Thatte HS, Anderson KC, and Munshi NC
- Subjects
- CD4 Lymphocyte Count, Case-Control Studies, Cell Proliferation, Humans, Immunity, Cellular, Lymphocyte Activation, Monoclonal Gammopathy of Undetermined Significance, T-Lymphocytes, Regulatory immunology, Multiple Myeloma immunology, T-Lymphocytes, Regulatory pathology
- Abstract
Multiple myeloma (MM) is characterized by the production of monoclonal immunoglobulin and is associated with suppressed uninvolved immunoglobulins and dysfunctional T-cell responses. The biologic basis of this dysfunction remains ill defined. Because T regulatory (T(reg)) cells play an important role in suppressing normal immune responses, we evaluated the potential role of T(reg) cells in immune dysfunction in MM. We observed a significant increase in CD4+ CD25+ T cells in patients with monoclonal gammopathy of undetermined significance (MGUS) and in patients with MM compared with healthy donors (25% and 26%, respectively, vs 14%); however, T(reg) cells as measured by FOXP3 expression are significantly decreased in patients with MGUS and MM compared with healthy donors. Moreover, even when they are added in higher proportions, T(reg) cells in patients with MM and MGUS are unable to suppress anti-CD3-mediated T-cell proliferation. This decreased number and function of T(reg) cells in MGUS and in MM may account, at least in part, for the nonspecific increase in CD4+ CD25+ T cells, thereby contributing to dysfunctional T-cell responses.
- Published
- 2006
- Full Text
- View/download PDF
50. A clinically relevant SCID-hu in vivo model of human multiple myeloma.
- Author
-
Tassone P, Neri P, Carrasco DR, Burger R, Goldmacher VS, Fram R, Munshi V, Shammas MA, Catley L, Jacob GS, Venuta S, Anderson KC, and Munshi NC
- Subjects
- Animals, Bone Transplantation, Cell Line, Tumor, Dexamethasone pharmacology, Disease Models, Animal, Fetal Tissue Transplantation, Green Fluorescent Proteins genetics, Humans, Immunotoxins pharmacology, Mice, Mice, SCID, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma immunology, Neoplasm Transplantation, Receptors, Interleukin-6 blood, Recombinant Proteins genetics, Spiro Compounds pharmacology, Transduction, Genetic, Transplantation, Heterologous, Multiple Myeloma pathology
- Abstract
We developed a novel in vivo multiple myeloma (MM) model by engrafting the interleukin 6 (IL-6)-dependent human MM cell line INA-6 into severe combined immunodeficiency (SCID) mice previously given implants of a human fetal bone chip (SCID-hu mice). INA-6 cells require either exogenous human IL-6 (huIL-6) or bone marrow stromal cells (BMSCs) to proliferate in vitro. In this model, we monitored the in vivo growth of INA-6 cells stably transduced with a green fluorescent protein (GFP) gene (INA-6GFP+ cells). INA-6 MM cells engrafted in SCID-hu mice but not in SCID mice that had not been given implants of human fetal bone. The level of soluble human IL-6 receptor (shuIL-6R) in murine serum and fluorescence imaging of host animals were sensitive indicators of tumor growth. Dexamethasone as well as experimental drugs, such as Atiprimod and B-B4-DM1, were used to confirm the utility of the model for evaluation of anti-MM agents. We report that this model is highly reproducible and allows for evaluation of investigational drugs targeting IL-6-dependent MM cells in the human bone marrow (huBM) milieu.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.